JVI Accepts, published online ahead of print on 4 December 2013 J. Virol. doi:10.1128/JVI.03183-13 Copyright © 2013, American Society for Microbiology. All Rights Reserved.

1

H3 stalk-based chimeric hemagglutinin influenza virus constructs protect mice

2

from H7N9 challenge

3

Florian Krammer1,#, Irina Margine1,2,#, Rong Hai1, Alexander Flood3, Ariana Hirsh1, Vadim Tsvetnitsky3,

4

Dexiang Chen3 and Peter Palese1,4*

5

1

6

2

7

3

8

4

9

*To whom correspondence should be addressed: [email protected]

10

Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA

Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA PATH, Seattle, WA, USA Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA

#Authors contributed equally

11 12

Abstract

13

The recent outbreak of H7N9 influenza virus infections in humans in China has raised concerns about the

14

pandemic potential of this strain. Here we test the efficacy of H3-stalk based chimeric hemagglutinin

15

universal influenza virus vaccine constructs to protect against H7N9 challenge in mice. Chimeric

16

hemagglutinin constructs protected from viral challenge in the context of different administration

17

routes, and a generic oil-in-water adjuvant similar to formulations licensed for use in humans.

18

Main Text

19

In early spring 2013 the Chinese authorities reported the first human cases of avian H7N9 influenza virus

20

infections (1) and a total of 137 cases and 45 fatalities have been officially confirmed by the World

21

Health Organization by October 25th, 2013 (including 2 cases in October 2013) (2). Characterization of

22

viral isolates revealed that H7N9 binds to alpha-2.6 linked sialic acids and that it can replicate in the

23

mammalian upper respiratory tract, indicating it could be able to acquire sustained human-to-human

24

transmissibility (3-5). These findings, together with the fact that neuraminidase (NA) inhibitor resistant

25

mutants have arisen quickly in treated patients - without apparent fitness loss (6) - have generated

26

concerns for an H7N9 pandemic.

27

The H7 hemagglutinin (HA) falls phylogenetically within the group 2 influenza A HAs and shares

28

conserved epitopes in the membrane proximal stalk domain with other members of this group (H3, H4,

1

29

H10, H14 and H15 HAs). This region of the HA is, however, considered to be immunologically

30

subdominant. Antibodies against this subdominant region of the HA are generally not induced by regular

31

- H3 containing - seasonal trivalent inactivated vaccines (TIV) and only to low titers by natural H3N2

32

infection (7-9). However, monoclonal antibodies recognizing epitopes in the stalk domain have been

33

isolated from mice and humans, and exhibit broadly neutralizing activity against divergent group 2

34

influenza virus subtypes (10-13).

35

We have recently reported that a vaccination regimen based on chimeric HAs (cHA) can elevate the

36

immunogenicity of this domain and thus focus the antibody response towards conserved epitopes in the

37

HA. Importantly, elicited anti-HA stalk antibody titers were sufficient to protect against influenza virus

38

challenges in both mice and ferrets (13-17). Importantly, protection is limited to viruses that express an

39

HA belonging to the same HA group used for vaccination and cross-reactivity among HAs belonging to

40

different groups was also not observed (14, 17). Here we aim to evaluate whether the titers of cross-

41

reactive stalk antibodies elicited by a group 2 (H3) HA stalk-based vaccination regimen are sufficient to

42

confer protection against the novel H7N9 virus. Furthermore, to study the importance of mucosal

43

responses to protection, we applied an experimental set-up that induces both, mucosal and systemic

44

immunity and compared it to one that would only induce systemic immunity. We also compared two

45

adjuvants, polyI:C (PIC) -which we successfully used in combination with cHAs in animals before -, and a

46

generic oil-in-water (OIW) adjuvant (18). The latter is similar to adjuvants that have been licensed for

47

use in humans (18). Animals (N=10 per group, female 6-8 week old BALB/c mice) were primed with a

48

DNA plasmid expressing a cH4/3 protein (H4 head derived from A/duck/Czech/57 on top of an H3 stalk

49

domain derived from A/Perth/16/09) (17) via intramuscular electroporation with a TriGrid

50

electroporation device (Ichor Medical Systems) (Fig 1A). It is of note that DNA vaccination is not

51

essential for induction of broad protection by cHA vaccination regimens. Vaccination with just proteins

52

(no DNA) yielded results comparable to vaccination with DNA priming in earlier studies (19). Three

53

weeks post prime, animals received a recombinant cH5/3 protein (H5 head derived from

54

A/Vietnam/1203/04 on top of an H3 stalk derived from A/Perth/16/09) (17). Animals in one group

55

received 5 ug of cH5/3 protein intranasally (i.n.) and 5 ug intramuscularly (i.m.) adjuvanted with 5 ug of

56

PIC (high molecular weight, Invivogen) ('PIC i.n.+i.m.'). A second group only received the i.m. dose with

57

PIC (5 ug HA in total) ('PIC i.m.'), while a third group received 5 ug of cH5/3 protein with a generic OIW

58

based adjuvant ('OIW i.m.') (Fig 1A). All mice were boosted a second time three weeks later with full

59

length H3 protein using the same vaccination routes, adjuvants and immunogen amounts, respectively

60

(Fig 1A). All recombinant proteins used for vaccination were expressed in the baculovirus expression 2

61

system with a C-terminal T4 foldon trimerization domain and a hexahistidine tag to facilitate purification

62

(20). The OIW adjuvant (20 mM citrate, 0.5% polysorbate 80, pH 6.5, 0.5% span-85 (sorbitan trioleate),

63

4.3% squalene) was prepared as described before (18) (Fig 2). Positive controls (n=5) received one i.m.

64

vaccination of formalin-inactivated A/Shanghai/1/13 H7N9 (SH1, 6:2 reassortant with HA and NA

65

derived from A/Shanghai/1/13 and the internal genes from A/Puerto Rico/8/34) whole virus

66

preparation. Negative controls (n=4-5) received a mock DNA vaccination followed by two boosts with

67

bovine serum albumin (BSA) administered in the same amounts and route with the respective cHA

68

vaccination regimens they were compared to. Four weeks after the last immunization, animals were

69

bled and then challenged with 10 murine lethal doses 50 (mLD50) of SH1 virus. Weight loss was

70

monitored over a period of 14 days and mice that lost more than 20 percent of their initial body weight

71

were euthanized. Animals in the PIC i.n.+i.m. group lost an average of 10% of their initial body weight

72

(Fig 1B) as compared to 15% in the PIC i.m. group (Fig 1C), suggesting that the mucosal immunity at the

73

site of infection has a significant role in protection. Furthermore, the mice in the OIW i.m. group lost

74

only 12% of their initial body weight on average (Fig 1D), which is also reflected in the observed survival.

75

All PIC i.m.+ i.n. vaccinated animals survived the challenge (Fig 1E), compared to only 70% of the PIC i.m.

76

animals (Fig 1F). However, survival in the OIW i.m. group was 100% (Fig 1F), indicating a better

77

protective effect with the OIW adjuvant. To quantitatively assess the titers of H7 HA antibodies elicited

78

by the three different vaccination regimens, we performed enzyme linked immunosorbent assays

79

(ELISA) using a recombinant SH1 H7 HA as substrate. Since the cHA-vaccinated animals were not

80

exposed to an H7 head domain, we concluded that any observed reactivity to H7 would be

81

predominantly derived from cross-reactive anti-stalk antibodies. To exclude binding to the hexahistidine

82

tag or the trimerization domain, we used a recombinant SH1 H7 HA expressed with a GCN pII leucine

83

zipper trimerization domain and a strep-tag II (14, 21). The highest endpoint titers were detected in

84

serum collected from PIC i.n.+i.m. vaccinated animals, followed by the OIW i.m. animals (Fig 3A). Mice

85

that received the PIC i.m. vaccine had statistically significant lower titers (p=0.03) than the OIW i.m.

86

animals. This difference in endpoint titers correlates well with the differences in weight loss and

87

survival. The antibody isotype distribution can be strongly influenced by adjuvants and can have a high

88

impact on the protective efficiency of antibodies. However, we did not detect significant differences in

89

the isotype profile when we compared the three vaccination regimens (Fig 3B). This suggest that - in the

90

case of i.m. only vaccination - the antibody titer was the main correlate of protection. Sera from the

91

vaccine and control groups were also tested in a micro-neutralization assay but results for the cHA

92

vaccinated groups were negative, probably due to the limit of detection (1:20) of this assay (data not 3

93

shown). Since we observed that animals immunized via both routes showed lower morbidity than

94

animals that were only vaccinated intramuscularly, we can speculate that mucosal IgA antibodies

95

induced by the i.n. vaccination had a substantial contribution to protection. Differences in weight loss

96

between the PIC i.n.+i.m. and the PIC i.m. group were statistically significant on day 7 (p=0.0383) and

97

day 8 (p=0.0136) (unpaired t-test). However, it should be noted that these animals also received twice

98

as much antigen as the i.m. only animals. In conclusion we show that mice can be protected against the

99

novel H7N9 virus using a stalk-based immunization regimen with cHA constructs that do not possess and

100

H7 head domain. Furthermore, an oil-in-water based adjuvant similar to those licensed for use in

101

humans (18, 22) performed well with the cHA vaccine candidates, suggesting that this combination

102

could also be considered for testing in humans. An HA stalk based vaccine providing universal influenza

103

virus protection could replace strain specific seasonal vaccines and further enhance our preparedness

104

against potential pandemic influenza virus strains like H7N9.

105

Acknowledgements

106

We thank Chen Wang for excellent technical support. Florian Krammer was supported by an Erwin

107

Schrödinger fellowship (J3232) from the Austrian Science Fund (FWF). This work was partially supported

108

by a Centers for Excellence for Influenza Research and Surveillance (CEIRS) grant (HHSN26620070010C

109

to PP), the NIH program project grant 1P01AI097092-01A1 (PP) and by HHSN272200900032C (Innate

110

Immune Receptors and Adjuvant Discovery; to PP).

111

References

112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127

1.

2. 3.

4.

Gao R, Cao B, Hu Y, Feng Z, Wang D, Hu W, Chen J, Jie Z, Qiu H, Xu K, Xu X, Lu H, Zhu W, Gao Z, Xiang N, Shen Y, He Z, Gu Y, Zhang Z, Yang Y, Zhao X, Zhou L, Li X, Zou S, Zhang Y, Yang L, Guo J, Dong J, Li Q, Dong L, Zhu Y, Bai T, Wang S, Hao P, Yang W, Han J, Yu H, Li D, Gao GF, Wu G, Wang Y, Yuan Z, Shu Y. 2013. Human infection with a novel avian-origin influenza A (H7N9) virus. N Engl J Med 368:1888-1897. http://www.who.int/influenza/human_animal_interface/influenza_h7n9/Data_Reports/en/. Ramos I, Krammer F, Hai R, Aguilera D, Bernal-Rubio D, Steel J, García-Sastre A, FernandezSesma A. 2013. H7N9 influenza viruses interact preferentially with 2,3-linked sialic acids and bind weakly to 2,6-linked sialic acids. J Gen Virol 94:2417-2423. Watanabe T, Kiso M, Fukuyama S, Nakajima N, Imai M, Yamada S, Murakami S, Yamayoshi S, Iwatsuki-Horimoto K, Sakoda Y, Takashita E, McBride R, Noda T, Hatta M, Imai H, Zhao D, Kishida N, Shirakura M, de Vries RP, Shichinohe S, Okamatsu M, Tamura T, Tomita Y, Fujimoto N, Goto K, Katsura H, Kawakami E, Ishikawa I, Watanabe S, Ito M, Sakai-Tagawa Y, Sugita Y, Uraki R, Yamaji R, Eisfeld AJ, Zhong G, Fan S, Ping J, Maher EA, Hanson A, Uchida Y, Saito T, Ozawa M, Neumann G, Kida H, Odagiri T, Paulson JC, Hasegawa H, Tashiro M, Kawaoka Y. 2013. Characterization of H7N9 influenza A viruses isolated from humans. Nature 501:551-555.

4

128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175

5.

6.

7.

8.

9.

10.

11.

12.

13. 14. 15.

16.

17.

18.

Xiong X, Martin SR, Haire LF, Wharton SA, Daniels RS, Bennett MS, McCauley JW, Collins PJ, Walker PA, Skehel JJ, Gamblin SJ. 2013. Receptor binding by an H7N9 influenza virus from humans. Nature 499:496-499. Hu Y, Lu S, Song Z, Wang W, Hao P, Li J, Zhang X, Yen HL, Shi B, Li T, Guan W, Xu L, Liu Y, Wang S, Tian D, Zhu Z, He J, Huang K, Chen H, Zheng L, Li X, Ping J, Kang B, Xi X, Zha L, Li Y, Zhang Z, Peiris M, Yuan Z. 2013. Association between adverse clinical outcome in human disease caused by novel influenza A H7N9 virus and sustained viral shedding and emergence of antiviral resistance. Lancet 381:2273-2279. Moody MA, Zhang R, Walter EB, Woods CW, Ginsburg GS, McClain MT, Denny TN, Chen X, Munshaw S, Marshall DJ, Whitesides JF, Drinker MS, Amos JD, Gurley TC, Eudailey JA, Foulger A, DeRosa KR, Parks R, Meyerhoff RR, Yu JS, Kozink DM, Barefoot BE, Ramsburg EA, Khurana S, Golding H, Vandergrift NA, Alam SM, Tomaras GD, Kepler TB, Kelsoe G, Liao HX, Haynes BF. 2011. H3N2 influenza infection elicits more cross-reactive and less clonally expanded antihemagglutinin antibodies than influenza vaccination. PLoS One 6:e25797. Wrammert J, Smith K, Miller J, Langley WA, Kokko K, Larsen C, Zheng NY, Mays I, Garman L, Helms C, James J, Air GM, Capra JD, Ahmed R, Wilson PC. 2008. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature 453:667-671. Margine I, Hai R, Albrecht RA, Obermoser G, Harrod AC, Banchereau J, Palucka K, García-Sastre A, Palese P, Treanor JJ, Krammer F. 2013. H3N2 influenza virus infection induces broadly reactive hemagglutinin stalk antibodies in humans and mice. J Virol 87:4728-4737. Wang TT, Tan GS, Hai R, Pica N, Petersen E, Moran TM, Palese P. 2010. Broadly protective monoclonal antibodies against H3 influenza viruses following sequential immunization with different hemagglutinins. PLoS Pathog 6:e1000796. Ekiert DC, Friesen RH, Bhabha G, Kwaks T, Jongeneelen M, Yu W, Ophorst C, Cox F, Korse HJ, Brandenburg B, Vogels R, Brakenhoff JP, Kompier R, Koldijk MH, Cornelissen LA, Poon LL, Peiris M, Koudstaal W, Wilson IA, Goudsmit J. 2011. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science 333:843-850. Corti D, Voss J, Gamblin SJ, Codoni G, Macagno A, Jarrossay D, Vachieri SG, Pinna D, Minola A, Vanzetta F, Silacci C, Fernandez-Rodriguez BM, Agatic G, Bianchi S, Giacchetto-Sasselli I, Calder L, Sallusto F, Collins P, Haire LF, Temperton N, Langedijk JP, Skehel JJ, Lanzavecchia A. 2011. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science 333:850-856. Krammer F, Palese P. 2013. Influenza virus hemagglutinin stalk-based antibodies and vaccines. Curr Opin Virol 3:521-530. Krammer F, Pica N, Hai R, Margine I, Palese P. 2013. Chimeric hemagglutinin influenza virus vaccine constructs elicit broadly protective stalk-specific antibodies. J Virol 87:6542-6550. Krammer F, Hai R, Yondola M, Tan GS, Leyva-Grado V, Ryder A, Rose JK, Palese P, GarciaSastre A, Albrecht RA. Assessment of influenza virus hemagglutinin stalk based immunity in ferrets, submitted. Pica N, Hai R, Krammer F, Wang TT, Maamary J, Eggink D, Tan GS, Krause JC, Moran T, Stein CR, Banach D, Wrammert J, Belshe RB, García-Sastre A, Palese P. 2012. Hemagglutinin stalk antibodies elicited by the 2009 pandemic influenza virus as a mechanism for the extinction of seasonal H1N1 viruses. Proc Natl Acad Sci U S A 109:2573-2578. Margine I, Krammer F, Hai R, Heaton NS, Tan GS, Andrews SA, Runstadler JA, Wilson PC, Albrecht RA, García-Sastre A, Palese P. 2013. Hemagglutinin Stalk-Based Universal Vaccine Constructs Protect against Group 2 Influenza A Viruses. J Virol 87:10435-10446. Ott G, Radhakrishnan R, Fang J-H, Hora M. 2000. The Adjuvant MF59: A 10-Year Perspective, p. 211-228. In O'Hagan DT (ed.), Vaccine Adjuvants, vol. 42. Springer. 5

176 177 178 179 180 181 182 183 184 185

19.

20.

21. 22.

Goff PH, Eggink D, Seibert CW, Hai R, Martínez-Gil L, Krammer F, Palese P. 2013. Adjuvants and immunization strategies to induce influenza virus hemagglutinin stalk antibodies. PLoS One 8:e79194. Krammer F, Margine I, Tan GS, Pica N, Krause JC, Palese P. 2012. A carboxy-terminal trimerization domain stabilizes conformational epitopes on the stalk domain of soluble recombinant hemagglutinin substrates. PLoS One 7:e43603. Weldon WC, Wang BZ, Martin MP, Koutsonanos DG, Skountzou I, Compans RW. 2010. Enhanced immunogenicity of stabilized trimeric soluble influenza hemagglutinin. PLoS One 5. O'Hagan DT, Ott GS, Nest GV, Rappuoli R, Giudice GD. 2013. The history of MF59(®) adjuvant: a phoenix that arose from the ashes. Expert Rev Vaccines 12:13-30.

186 187

Figure Legends

188 189 190 191 192 193 194 195 196 197 198 199 200

Figure 1: Vaccination with H3 stalk-based cHA protects mice from challenge with H7N9 virus. A. Schematic representation of the vaccination regimen. Structures are based on protein database structure # 1RU7. Mice in B-G received a DNA prime coding for cH4/3 protein (H4 head in combination with and H3 stalk) and were then boosted with cH5/3 (H5 head on top of an H3 stalk) and full length H3 protein. The animals were challenged four weeks after the last immunization with H7N9 virus. Weightloss curves are shown in B-D, survival curves are shown in E-G. The dashed line in C and D indicates the highest average weight loss (day 7) of the 'PIC i.n. + i.m.' group shown in B. Mice shown in B and E (PIC i.n. +i.m.) were boosted sequentially with cH4/3 and cH5/3 protein intranasally and intramuscularly in the presence of polyI:C as adjuvant. Animals in C and F received the same proteins as boost but received the immunizations only intramuscularly (PIC i.m.). Mice in D and G were immunized intramuscularly only with an oil-in-water adjuvant (OIW i.m.). Animals in the control groups received BSA with the respective adjuvant and via the respective routes. Positive control animals (same group was compared to all three experimental conditions) were vaccinated intramuscularly with inactivated matched challenge virus.

201 202 203 204 205 206 207 208 209 210 211

Figure 2: Oil-in-water (OIW) adjuvant preparation and characteristics. A Scheme of the OIW adjuvant preparation. The OIW emulsion is prepared at 20-40 mL scale. Span-85 is dissolved in squalene, polysorbate 80 is dissolved deionized water and then mixed with citrate-citric acid buffer. Oil and aqueous phase fractions are combined and mixed into a coarse emulsion by vortexing which is then passed through an LV1 Microfluidizer (Microfluidics, Westwood, MA) at 12,000 psi. The eluent is collected and re-passaged for a total of 5 passes through the microfluidizer to yield a stable and homogeneous emulsion. The final pass is filtered through 0.22µm filter and filled into sterile glass vials. B Particle size of the OIW adjuvant prepared by microfluidization was determined by dynamic light scattering on a Malvern Nano 3ZS. The sizes shown are the Z-average mean from 2 independently prepared batches. These preparations are considered monomodal as polydispersity after the final passage through the microfluidizer and sterile filtration ranged from 8-11%.

212 213 214

Figure 3: Vaccination with H3 stalk-based cHAs induces high antibody titers against H7 hemagglutinin in mice. A IgG endpoint titers of cHA vaccinated mice against H7 (A/Shanghai/1/13) HA. The one-tailed p value was calculated using an unpaired t test. B Immunoglobulin isotype distribution directed to H7 HA

6

215 216

in sera from mice vaccinated via different routes and with different adjuvants. The analysis was performed at a 1:200 dilution.

7

H3 stalk-based chimeric hemagglutinin influenza virus constructs protect mice from H7N9 challenge.

The recent outbreak of H7N9 influenza virus infections in humans in China has raised concerns about the pandemic potential of this strain. Here, we te...
1MB Sizes 0 Downloads 0 Views